Review

Cellular mechanisms underlying obesity-induced arterial stiffness

Published Online:https://doi.org/10.1152/ajpregu.00235.2016

Abstract

Obesity is an emerging pandemic driven by consumption of a diet rich in fat and highly refined carbohydrates (a Western diet) and a sedentary lifestyle in both children and adults. There is mounting evidence that arterial stiffness in obesity is an independent and strong predictor of cardiovascular disease (CVD), cognitive functional decline, and chronic kidney disease. Cardiovascular stiffness is a precursor to atherosclerosis, systolic hypertension, cardiac diastolic dysfunction, and impairment of coronary and cerebral flow. Moreover, premenopausal women lose the CVD protection normally afforded to them in the setting of obesity, insulin resistance, and diabetes, and this loss of CVD protection is inextricably linked to an increased propensity for arterial stiffness. Stiffness of endothelial and vascular smooth muscle cells, extracellular matrix remodeling, perivascular adipose tissue inflammation, and immune cell dysfunction contribute to the development of arterial stiffness in obesity. Enhanced endothelial cortical stiffness decreases endothelial generation of nitric oxide, and increased oxidative stress promotes destruction of nitric oxide. Our research over the past 5 years has underscored an important role of increased aldosterone and vascular mineralocorticoid receptor activation in driving development of cardiovascular stiffness, especially in females consuming a Western diet. In this review the cellular mechanisms of obesity-associated arterial stiffness are highlighted.

INTRODUCTION

Cardiovascular (CV) disease (CVD) is the leading cause of death not only in the United States and but also worldwide (3, 30). Arterial stiffness is an independent risk factor that contributes to the development and progression of CVD as evidenced by epidemiological studies (5, 129). The prevalence of obesity in the United States and worldwide is increasing in epidemic proportions, leading to increases in the incidence of arterial stiffness and associated CVD (87, 121, 122). The driving forces that contribute to obesity-associated arterial stiffness are increased consumption of a diet rich in saturated fat and refined carbohydrates [i.e., a Western diet (WD)] and a sedentary lifestyle (5, 23, 25, 138). Furthermore, the obesity-associated development of vascular stiffness is more common in women than men, and obese and insulin-resistant women lose the CVD protection normally afforded to them (9, 82) (Fig. 1).

Fig. 1.

Fig. 1.Causes and consequences of obesity-associated arterial stiffness. RAAS, renin-angiotensin-aldosterone system; SNS, sympathetic nervous system; M1 and M2, macrophages; IL-10, interleukin 10; Treg, regulatory T cell; Th1 and Th17, T helper 1 and 17 cells; ER, endoplasmic reticulum; eNOS, endothelial nitric oxide synthase; PAI-1, plasminogen activator inhibitor 1; TPA, tissue plasminogen activator; CVD, cardiovascular disease; CKD, chronic kidney disease.


Arterial stiffness is associated with components of cardiorenal metabolic syndrome (CRS), a constellation of interactive cardiac and metabolic risk factors, including overweight/obesity, hypertension, insulin resistance/hyperinsulinemia, metabolic dyslipidemia, and microalbuminuria/impaired renal function (6, 103) (Fig. 1). Although arterial stiffness increases with cardiovascular aging, the process is accelerated and occurs earlier in the presence of obesity, insulin resistance, and diabetes (54, 74). Therefore, prevention and treatment directed to attenuation of arterial stiffness are emerging as important targets for improved outcomes for CVD and chronic kidney disease (CKD) in those with obesity and diabetes (9, 53). Development of arterial stiffness is a complex process that is driven by the interaction of endocrine factors and cytokines, as well as interactions between different vascular cellular components, the extracellular matrix (ECM), perivascular adipose tissue (PVAT), and immune cells in the vasculature (5, 19, 53). In this review we focus on cell-specific mechanisms that promote arterial stiffening (Fig. 2), including the role of diet-induced obesity-related abnormalities of the endothelium as a critical determinant of vascular stiffening. This review includes a discussion of the role of endothelial abnormalities in the promotion of impaired endothelial nitric oxide (NO) synthase (eNOS) activation and associated increases in vascular stiffness. We also discuss the role of vasoactive factors in promoting arterial stiffness, the emerging role of cell-specific mineralocorticoid receptor (MR) activation in promoting endothelial cortical stiffness via endothelial Na+ channel (EnNaC) activation, and the impact of a decrease in bioavailable NO. We also address the concept that a reduction of bioavailable NO leads to activation of transglutaminase 2 (TG2), a collagen cross-linking enzyme, thereby promoting vascular fibrosis. We further describe the role of maladaptive immune inflammatory responses and oxidative stress in the development of endothelial dysfunction and arterial stiffening. In addition, we briefly review the emerging role of PVAT in promoting arterial stiffening. We then address a vulnerable population, namely, obese/diabetic females, who are susceptible to diet-induced obesity-mediated arterial stiffening and associated increases in CVD and CKD.

Fig. 2.

Fig. 2.Endothelial cell (EC)-macrophage-vascular smooth muscle cell interactions in obesity-associated arterial stiffness. Impaired insulin metabolic signaling in ECs leads to decreased activation of endothelial nitric oxide (NO) synthase (eNOS) and NO bioavailability. Increase in endothelial Na+ channel (EnNaC) activity occurs due to mineralocorticoid receptor (MR)-induced activation of serum/glucocorticoid regulated kinase 1 (SGK1) and increased EnNaC expression, which result in increased influx of Na+ and polymerization of F actin, leading to endothelial cortical stiffness. This leads to impaired flow-mediated release of NO. Increased oxidative stress due to RAAS-induced activation of NADPH oxidase, xanthine oxidase, and mitochondrial oxidative stress results in increased destruction of NO. eNOS uncoupling leads to further decreases in bioavailable NO. Decreased bioavailable NO causes increased expression of adhesion molecules on ECs, favoring monocyte recruitment. Decreased bioavailable NO also results in macrophage (MΦ) activation, M1 macrophage polarization, and oxidative stress. This leads to further destruction of NO. Decreased bioavailable NO results in transglutaminase 2 (TG2) activation, which promotes collagen cross-linking and causes cortical stiffness, which impairs flow-mediated NO production. Decreased heme oxygense 1 (HO-1) activity further contributes to the decrease in bioavailable NO, thereby promoting arterial stiffness. ECM, extracellular matrix; ERK1/2, extracellular-regulated kinase 1/2; IRS-1, insulin receptor substrate-1; PI3K, phosphatidylinositol 3-kinase; ROS, reactive oxygen species; S6K1, S6 kinase 1.


ARTERIAL STIFFNESS, CRS, AND PROGRESSION OF CVD AND CKD: CHICKEN-AND-EGG RELATIONSHIP

The relationship between arterial stiffness and the development of CRS and the progression of CVD and CKD appears to be a chicken-and-egg dilemma (7, 122). Arterial stiffness in obese children often precedes the development of hypertension, suggesting that arterial stiffness is one of the earliest biomarkers for increased CVD risk (36). Although hypertension is considered an important risk factor for the development of vascular remodeling and stiffness as an adaptive process, a cause-and-effect relationship exists between hypertension, especially systolic hypertension, and aortic stiffness (92). Arterial stiffness is often increased in overweight/obese humans and animal models before development of hypertension (35). Based on the magnitude of the increase in arterial stiffness, an incremental increase occurs in the development of hypertension (74). Moreover, rodent studies show that arterial stiffness precedes hypertension in mice fed a translational high-fat/high-fructose diet (i.e., WD) and that removal of this diet results in decreased arterial stiffness (139).

Increased arterial stiffness is significantly associated with cardiovascular and renal dysfunction, as well as CVD and CKD (2022, 39, 54, 86). Increased systolic pressure and decreased diastolic pressure, resulting in increased pulse pressure, are caused by stiffening of the central arteries. An increase in systolic pressure increases cardiac afterload, left ventricular mass, and oxygen demand, whereas a decrease in diastolic pressure impairs coronary blood flow. These changes can result in left ventricular remodeling, ischemia, and fibrosis, all of which contribute to left ventricular diastolic dysfunction and coronary heart disease (16, 57, 83). However, development of diastolic dysfunction also occurs independent of alterations in blood pressure, perhaps driven by endothelial stiffness and dysfunction, which promote cardiomyocyte stiffness and cardiac fibrosis (8, 58). Arterial stiffness promotes kidney disease, and an increase in arterial stiffness is also considered a promoter of CVD in patients with CKD (20, 39, 137).

MEASUREMENT OF ARTERIAL STIFFNESS

Noninvasively, in vivo measurement of arterial stiffness is usually accomplished by 1) ultrasound measurement of arterial compliance and distensibility, 2) measurement of the velocity of the pressure wave traveling between two segments of an artery to determine pulse wave velocity (PWV), and 3) measurement of augmentation pressure divided by blood pressure to determine the augmentation index (108). PWV is considered an index for arterial wall stiffness, whereas the augmentation index is related to both arterial wall stiffness and wave reflections, which in turn are dependent on variations in heart rate and peripheral resistance (53). PWV measurement is considered the gold standard for evaluation of vascular stiffness (53, 80). PWV in animal models is usually determined by measurement of the pressure wave at the aortic arch and downstream in the lower abdominal aorta (26). Ex vivo measurements of arterial stiffness are often accomplished by uniaxial or biaxial loading of arterial segments and study of pressure-diameter and force-length behavior (26, 135). The use of atomic force microscopy (AFM) greatly enhances the sensitivity for determination of endothelial vascular smooth muscle cell (VSMC) and cardiomyocyte stiffness ex vivo and in vitro (58, 116). AFM, in combination with fluorescence imaging, is used to simultaneously measure vascular cell stiffness and cytoskeletal dynamics. A novel method combining AFM and confocal imaging has been used to examine cortical actin reorganization in VSMCs (114, 115).

ARTERIAL FUNCTIONAL AND STRUCTURAL ABNORMALITIES PROMOTING OBESITY-ASSOCIATED ARTERIAL STIFFNESS: ROLE OF CELLULAR AND ECM INTERACTIONS PROMOTING ARTERIAL STIFFNESS IN DIET-INDUCED OBESITY

Arterial stiffness in obesity is associated with structural and functional changes in the intimal, medial, and adventitial layers of the vasculature (119). The endothelium is a component of the arterial intima, which is separated from the media by the internal elastic lamina. The medial layer in larger conduit vessels consists of elastic lamina that are arranged as concentric layers and interspersed with smooth muscle cells and collagen (78, 123). The adventitial layer is enriched with fibroblasts, collagen, immune cells, and adipose tissue (123). Arterial stiffness is regulated by plasma-derived factors, as well as factors derived from the different layers of the vascular wall. Moreover, interactive signaling between different cells of the vascular wall modulates structure and function of cellular and noncellular components (53, 75, 110a, 149). Intimal thickening is a complex process that is associated with endothelial dysfunction and fibrotic remodeling. Although ECM remodeling due to quantitative and qualitative changes in elastin and collagen has been extensively studied, recent studies demonstrate new regulators, including matrix metalloproteinases (MMP), in modulation of the ECM (75). However, increased arterial stiffness in insulin-resistant and prediabetic states has been related to thickness of the vascular wall, suggesting that mechanisms related to vascular cells that are different from ECM remodeling are also critical in determining vascular stiffness. In addition to ECM remodeling via changes in elastin and collagen, recent studies demonstrate a role for both endothelial and vascular smooth muscle stiffness, leading to the use of such terms as “stiff endothelial cell syndrome” (68) and “smooth muscle stiffness syndrome” (115). In addition to the role of endothelial cells (ECs) and VSMCs, vascular adipose and immune cell dysfunction contribute significantly to obesity-associated arterial stiffness (19, 29, 37, 134), thereby underscoring the importance of cellular and ECM interactions in obesity-associated arterial stiffness (53, 82).

VASOACTIVE FACTORS PROMOTING OBESITY-ASSOCIATED ARTERIAL STIFFNESS

Hyperinsulinemia and Inappropriate Activation of the Renin-Angiotensin-Aldosterone System

Insulin resistance associated with hyperinsulinemia precedes the development of CVD and CKD with obesity and the CRS (14, 93, 117). This view is supported by studies demonstrating reduction of the vasorelaxation responses to insulin, but not acetylcholine, before the onset of hypertension in aged rats (112) and rats developing spontaneous hypertension (73). Systemic and CV insulin resistance and related arterial stiffness in the setting of obesity are, in large part, due to the interactions between diet and activation of the systemic, as well as tissue, renin-angiotensin-aldosterone system (RAAS) (67, 94). Increased production of angiotensin II (ANG II) and uric acid in the vasculature as a result of a high-fructose diet underscores the importance of activation of the tissue RAAS and hyperuricemia in promotion of CV remodeling and stiffness (5). ANG II and aldosterone act in concert to promote CV remodeling and stiffness in states of insulin resistance and obesity (14, 107). The critical role of activation of cell-specific vascular MRs in vascular remodeling and stiffening in states of diet-induced obesity, especially in females, is highlighted by recent studies (53, 56, 103). Patients with primary hyperaldosteronism have insulin resistance, and plasma aldosterone elevations are associated with obesity and insulin resistance in normotensive subjects (14, 56, 141). Multiple studies, including those conducted by our laboratory using different models of obesity, have shown that treatment with MR antagonists improves CV function (5, 11, 26, 66). Endothelin-1, the expression of which is increased by RAAS activation, is one of the instigators of increases in vascular stiffness and remodeling (94, 95).

Sympathetic Nervous System Activation

Obesity is often associated with increased sympathetic nervous system (SNS) activity, which is a key factor contributing to arterial stiffness and hypertension (25, 121). Hyperinsulinemia, hyperleptinemia, inappropriate activation of systemic and tissue RAAS, baroreflex dysfunction, and obstructive sleep apnea in obesity are associated with inappropriate activation of the SNS (9, 25). The precise mechanisms by which enhanced SNS activity contributes to arterial stiffness remain to be determined.

Adipocyte-Derived Factors

Metabolic changes in adipose tissue in the setting of obesity result in altered secretion of bioactive molecules and hormones, including tumor necrosis factor-α (TNF-α), interleukin (IL)-6, angiotensinogen, aldosterone-stimulating factors, aldosterone, adiponectin, dipeptidylpeptidase 4 (DPP-4), leptin, resistin, and monocyte chemoattractant protein-1 (MCP-1) (43, 99, 108). Elevated circulating levels of adipocyte-derived inflammatory cytokines may impair vascular insulin sensitivity and increase recruitment and activation of proinflammatory immune cells in the vasculature, which then contribute to development of arterial stiffness. Moreover, greater lipolytic activity in visceral adipose tissue in insulin-resistant states leads to increases in free fatty acids. Free fatty acids inhibit insulin-stimulated glucose uptake and metabolic insulin signaling in adipose, as well as vascular, tissue (25, 53). Decreased insulin metabolic signaling in ECs leads to reductions in NO production, which in turn, promote stiffening of ECs and, subsequently, vascular stiffening.

High-Fructose Diet

Increased serum uric acid levels are frequently seen in patients with obesity, insulin resistance, hypertension, CVD, and CKD (20, 55). An increase in uric acid occurs with excessive consumption of a WD that is rich in fructose (104). Indeed, fructose promotes hepatic production of uric acid (6). Our research group has reported increased cardiac xanthine oxidase activity and oxidative stress in WD-fed mice (57). Furthermore, we have observed that inhibition of tissue xanthine oxidase by allopurinol decreases vascular stiffness and prevents maladaptive immune and inflammatory responses in the vasculature (6, 27, 70). Increased expression of xanthine oxidase occurs in concert with increased RAAS activation and oxidative stress, which collectively lead to increased arterial stiffness in diet-induced obesity (20, 104).

CELLULAR MECHANSIMS OF ARTERIAL STIFFNESS

Endothelium

Decreased NO production by impaired insulin metabolic signaling and increased oxidative stress-mediated destruction of NO lead to reduced bioavailable NO.

Insulin metabolic signaling is critical for regulation of normal endothelial function and suppression of vascular stiffness (Fig. 2). In vascular ECs, insulin metabolic signaling promotes eNOS activation through an insulin receptor substrate (IRS)-1/phosphatidylinositol 3-kinase (PI3K) signaling/protein kinase B (Akt)-mediated pathway (Fig. 2) (54). On the other hand, insulin-mediated growth pathway signaling results in activation of extracellular-regulated kinase 1/2 and endothelin-1 production. Insulin metabolic signaling in ECs and VSMCs is inhibited by ANG II and aldosterone, resulting in impairment of downstream antioxidant effects and the anti-inflammatory response of the insulin metabolic signaling pathway (8). This, in turn, leads to impaired NO-mediated vasodilation responses to insulin, and decreases in NO lead to increased arterial stiffness. The mechanisms underlying RAAS-mediated insulin resistance in ECs and VSMCs are not well understood. One important signaling pathway that leads to impaired insulin metabolic signaling involves increased serine and impaired tyrosine phosphorylation of IRS-1, which is promoted by activation of kinases, including protein kinase C and p70 S6 kinase 1 (p70S6K1) (7, 8). ANG II-mediated increases in serine phosphorylation of IRS-1 through activation of the mammalian target of rapamycin/S6K1 signaling pathway result in impairment of IRS-1/2/PI3K/Akt signaling and decreased phosphorylation/activation of eNOS in ECs (65). Recently, we examined interactions between cell-specific MR activation and associated impairment of insulin metabolic signaling. Insulin metabolic signaling was compromised by impairment of Akt phosphorylation and decreased phosphorylation/activation of eNOS in WD-fed female mice. Inhibition of vascular MR activation by low-dose spironolactone or deletion of MRs in ECs prevented a decrease in eNOS phosphorylation/activation and decreases in bioavailable NO in the setting of diet-induced obesity (26, 56).

Impairment of flow-mediated vasodilation and decreased bioavailable NO: emerging role of the EnNaC in endothelial stiffness and associated inhibition of flow-mediated NO release.

Laminar blood flow-generated shear stress is an important regulator of eNOS activation and vasodilation (60). Impaired flow-mediated dilation is seen in obesity-associated arterial stiffness, which, in turn, is associated with CV insulin resistance (56). An increase in fluid shear stress normally activates IRS-1/PI3K/Akt signaling, leading to phosphorylation of eNOS. This process depends on normal structure and function of the endothelial glycocalyx and its interactions with the cytoskeleton (77, 145). Recent studies have provided new insights into the regulation of flow-mediated release of NO through interaction of the glycocalyx and the EnNaC in comparison with the epithelial Na+ channel (ENaC, in renal tubular epithelial cells) in the EC plasma membranes (34, 52). ENaC consists of three subunits, α, β, and γ: the α-subunit is essential for proper function of ENaC, while the β- and γ-subunits act as amplifiers (34). Studies on mesenteric arteries have demonstrated improvement of flow-mediated dilation in mesenteric arteries by inhibition of EnNaC activation, which is associated with increased Akt phosphorylation and eNOS activation (102).

MR activation of the EnNaC and endothelial stiffness: endothelial salt sensitivity.

Classically, activated MRs in distal renal tubular cells bind the ENaC promoter, inducing ENaC expression (52, 56). MR activation in distal renal tubular cells also mediates aldosterone-induced mammalian target of rapamycin/serum/glucocorticoid-regulated kinase 1 activation, which, in turn, causes increased accumulation of ENaC at the plasma membrane. Recent studies have demonstrated MR regulation of EnNaC. EC MR activation increases abundance of the EnNaC at the membrane, facilitating Na+ entry, triggering G-actin polymerization to F-actin. This results in stiffening of the plasma membrane and the immediate submembrane compartment and decreased caveolar release and activation of eNOS (28). We recently reported increased expression of vascular MR and EnNaC in the aorta associated with increased endothelial and arterial stiffness in WD-fed female mice. However, deletion of MR selectively in ECs resulted in decreased EnNaC activity and increased bioavailable NO, which lead to decreased arterial stiffness (56). Moreover, administration of a very low dose of the EnNaC inhibitor amiloride also improved endothelial function and reduced vascular stiffness in vivo and improved flow-mediated dilation of arteries in vitro (56, 84). These data suggest an important role for EC MRs and vascular EnNaC activation in promotion of obesity-associated arterial stiffness.

Decreased bioavailable NO is also mediated through reactive oxygen species-mediated NO inactivation.

Inactivation of NO by reactive oxygen species (ROS) is another mechanism responsible for reduced NO bioavailability in insulin-resistant states (8). In addition to sources of ROS contributed by mitochondrial activation of NADPH oxidase and xanthine oxidase, uncoupling eNOS can also generate superoxide anions (O2) in conditions of inadequate l-arginine or tetrahydrobiopterin availability (7). Accumulating evidence suggests that angiotensin type 1 receptor and MR activation stimulates vascular NADPH oxidase activity and ROS generation. This increase in ROS, in turn, results in reduced bioavailability of NO by its conversion to peroxynitrite and by uncoupling eNOS to generate more ROS in a positive-feedback loop in ECs (65). Recently, we showed increased production of 3-nitrotyrosine, an index of oxidative stress, and reduced bioavailable NO in WD-fed female mice (26). Suppression of 3-nitrotyrosine accumulation by either MR antagonism using low-dose spironolactone (26) or deletion of MR in ECs (56) resulted in improvement of arterial stiffness. These observations further support the view that EC-mediated ROS accumulation is an important factor leading to reduced bioavailable NO. ROS production by xanthine oxidase activation in the vasculature in obesity is another mechanism leading to destruction of NO (20), and inhibition of xanthine oxidase increases bioavailable NO and decreases arterial stiffness (27). Mitochondria are also important sources of endothelial generation of ROS (53), and increased vascular ROS compromises insulin metabolic signaling and associated eNOS activation, thereby lessening bioavailable NO and promoting vascular stiffness. The increase in endoplasmic reticulum (ER) stress in obesity also contributes to generation of ROS, reductions in bioavailable NO, and increases in vascular stiffness (7).

Reduced heme oxygenase-1, reduced bioavailable NO, and arterial stiffness.

Breakdown of heme catalyzed by heme oxygenase-1 (HO-1) results in the production of carbon monoxide, biliverdin, and ferrous iron. Biliverdin is converted to bilirubin. The beneficial effects of HO-1 occur through increases in carbon monoxide and bilirubin (1, 32). HO-1 activation stimulates eNOS expression and inhibits NADPH oxidase and xanthine oxidase expression, thereby reducing ROS and enhancing NO signaling bioavailability (1, 50, 120). Induction of HO-1 reduces oxidative stress, increases bioavailable NO, and improves insulin metabolic signaling, which is associated with rodent models of obesity (1, 48).

Immune Cells

Maladaptive immune and inflammatory responses.

The role of immune and inflammatory responses in the development of arterial stiffness is increasingly recognized (3, 9, 112, 113). Inflammatory M1 macrophage activation is associated with increased production of ROS and reactive nitrogen species. Promotion of T helper 1 (Th1) responses (62) and increased expression of proinflammatory cytokines, such as TNF-α, IL-6, and MCP-1, are also associated with M1 macrophage activation. The proinflammatory cytokines, such as TNF-α, IL-1, and IL-6, reduce vascular insulin metabolic signaling. In contrast, alternatively activated M2 macrophages, which express YM1, arginase-1, and IL-10, decrease inflammation. Increased tissue infiltration of M1 macrophages associated with accumulation of ROS and development of insulin resistance is also enhanced by Th1 cells and cytotoxic CD8+ T cells. On the other hand, inflammatory T cells are suppressed by T lymphocyte regulatory (Treg) cells (63). Treg cell accumulation is decreased in adipose tissue from mice fed a high-fat diet (9). Treg cells normally improve insulin sensitivity, in part by inhibition of M1 macrophage polarization, as well as M2 macrophage-mediated secretion of IL-10. This process corrects impaired insulin metabolic signaling caused by proinflammatory cytokines and inhibits NADPH oxidase and reduces ROS generation in ECs (63). IL-17 is a cytokine secreted by T helper 17 (Th17) cells. Th17 cells are elevated in obese and type 2 diabetes mellitus patients as well as obese mice (9, 63). Increased IL-17 levels are associated with decreased insulin sensitivity and insulin resistance, whereas suppression of IL-17 levels results in increased insulin sensitivity (148). These immune mechanisms that contribute to impaired insulin metabolic signaling are also associated with immune inflammatory responses that promote arterial stiffness (25, 42, 112, 140).

EC-immune cell interactions in macrophage recruitment and activation.

Recruitment of immune cells to vascular and adipose tissue occurs early during the development of vascular dysfunction and arterial stiffness (9). Immune cell recruitment is a sequential, multistep event resulting from interaction of monocytes and EC adhesion molecules (105, 111) (Fig. 2). Endothelial inflammation and oxidative stress appear to be critical in the recruitment of immune cells, including monocytes and macrophages, to ECs (51, 53). The significance of EC-dependent macrophage recruitment is underscored by significant inhibition of macrophage recruitment to the heart in EC MR knockout (KO) mice (58, 146). In this regard, EC MR activation leads to increased expression of adhesion molecules, as well as expression of MCP-1, which promotes attraction and adhesion of monocytes to ECs (15, 58). Both decreased availability of NO and increased accumulation of ROS increase the expression of EC adhesion molecules and MCP-1 (79). Decreased bioavailable NO also stimulates the M1 macrophage proinflammatory response once the macrophages have infiltrated the vasculature (71).

Inflammasomes.

Inflammasome activation also contributes to promotion of the inflammatory response, insulin resistance, type 2 diabetes, and vascular stiffness through increases in IL-1β (46). Exposure to pathogens or activation of danger-associated signals in inflammatory states results in inflammasome activation (106). In obesity, vascular inflammasomes are activated by elevated tissue levels of palmitate and ceramide (46, 106). Uric acid also activates inflammasomes, and the improvement seen after allopurinol administration may, in part, be caused by suppression of this proinflammatory response (20, 104).

Vascular Smooth Muscle Cells

Recent studies demonstrate that intrinsic VSMC alterations also contribute to arterial stiffness (115). VSMC stiffness increases in response to vasoactive agonists associated with adhesion to ECM proteins (47, 115). Recent studies favor the role of VSMC MRs in vascular stiffness. Deletion of MRs, specifically in VSMCs, decreases accumulation of ROS in the vasculature and is associated with decreased vasoconstriction in response to agonists (88). In addition, deletion of smooth muscle cell MRs prevents an aldosterone/salt-induced increase in arterial stiffness (40). However, the mechanisms regulating the passive stiffness of VSMCs and increased adhesive properties of VSMCs to ECM are not clearly known, and further studies will define the role of MRs in VSMC regulation of arterial stiffness in the setting of obesity.

Extracellular Matrix

Collagen accumulation and cross-linking of collagen.

ECM remodeling and fibrosis are important contributors to arterial stiffness (53). The ECM is composed of collagen, elastin, and specialized proteins, such as fibronectin and proteoglycans. Extracellular remodeling includes changes in accumulation of ECM material as well as qualitative changes in the ECM. Accumulation of vascular collagen is seen in obesity-associated arterial stiffness. One of the underlying mechanisms for this process is enhanced transforming growth factor-β/SMAD signaling and increased connective tissue growth factor expression (26, 53). We recently showed that vascular MR expression and activation are enhanced with increased aortic fibrosis in female WD-fed mice (26, 56). In addition to collagen accumulation, increased cross-linking is mediated by increased activation of TG2 and/or lysyl oxidase or by nonenzymatic glycation of collagen by advanced glycation end products (AGEs). In mice fed a high-fat–high-refined carbohydrate WD, arterial stiffness is associated with altered collagen structure, such as cross-linking of collagen by TG2 activation (132). TG2 catalyzes formation of ε (γ-glutamyl) lysine isopeptide, in a Ca2+-dependent manner (132). The substrates for TG2 include collagen, fibronectin, laminin, and actin (139). Endothelial glycocalyx polymerization involves increased accumulation of F actin, which, in turn, contributes to increased endothelial cortical stiffness and associated reductions in eNOS activation and reduced bioavailable NO (38, 39). Decreased bioavailable NO, in turn, promotes TG2 activation (33). Transforming growth factor-β has also been shown to increase TG2 expression and activation (132). Activation of TG2 is regulated by the presence of Ca2+ as well as nitrosylation (61). Nitrosylation promoted by NO decreases TG2 activity and inhibits release of TG2 from ECs (61). Therefore, a decrease in bioavailable NO, as seen in obesity, may significantly contribute to cross-linking of collagen by causing activation and extracellular release of TG2 (61). In this regard, increased TG2 activity and cross-linking of collagen with increased arterial stiffness have been demonstrated in mice fed a high-fat–high-refined carbohydrate diet (139). Cross-linking of collagen by lysyl oxidase also contributes to arterial stiffness in obesity (135). In addition to cross-linking of collagen, lysyl oxidase-derived hydrogen peroxide has also been shown to contribute to arterial stiffening through the p38 mitogen-activated protein kinase signaling pathway (85). Increased lysyl oxidase also causes abnormal elastin structure, which contributes to arterial stiffness (85).

Accumulation of AGEs due to consumption of a diet high in fructose increases collagen cross-linking, thereby promoting arterial stiffness (64, 97, 124). AGEs are formed by nonenzymatic glycation of proteins, lipids, and nucleic acids formed via a Maillard reaction. AGE accumulation occurs with aging, but the process is accelerated in obesity (124). However, AGE-induced effects on arterial stiffness are mediated by multiple mechanisms, including decreases in bioavailable NO and activation of the receptor for AGEs (10, 124). Obesity-mediated activation of the receptor for AGEs occurs by multiple mechanisms, including inappropriate activation of the RAAS and dysregulation of the DPP-4/incretin system (10, 143, 144).

Elastin breakdown.

Vascular elastin breakdown is also an important cause of arterial stiffness. Several MMPs and their endogenous inhibitors of metalloproteinases are implicated in the breakdown of elastin (49, 125). Among MMPs, MMP-2, MMP-9, and MMP-12 play an important role in ECM remodeling in the vasculature and arterial stiffness (12, 13, 26, 75). Serum levels of MMP-9 are increased in obese patients, and MMP-2 levels were increased in obese adults and children (42). Adiponectin has been shown to decrease MMP-2 activity, and increased MMP-2 activity in hypertensive obese children is associated with decreased adiponectin production (13). MMPs are secreted by ECs, VSMCs, macrophages, and T lymphocytes (49). In addition to their effects on elastin degradation, MMPs also induce proinflammatory responses that promote arterial stiffness (49, 91). We recently reported increased breakdown and distortion of elastic lamina in the aorta of female mice fed a WD. This same breakdown was attenuated in WD-fed EC MR KO mice, suggesting a role for EC-specific MR activation in modulating elastin breakdown (56).

Perivascular Adipose Tissue

Adipose tissue was originally considered a simple lipid rich in connective tissue, but the presence of immune cells, production of biologically active compounds, and specialized functions of adipocytes suggest the role of an endocrine tissue for adipose tissue. Recent studies demonstrated dynamic interconversion of brown and white adipose tissue with altered cytokine production and remodeling responses. This underscores the importance of adipose tissue as one of the therapeutic targets to reduce CVD (17, 44, 59, 96, 101). In this regard, adipose tissue surrounding the vasculature, PVAT, not only serves as a structural support for most arteries but also secretes molecules with paracrine effects on the vasculature (41, 127). PVAT secretory products include proinflammatory/anti-inflammatory molecules and vasoconstrictive/vascular relaxing factors, including TNF-α, IL-6, angiotensinogen, aldosterone-stimulating factors, aldosterone, adiponectin, DPP-4, leptin, resistin, and MCP-1 (43, 99, 108). Hyperplasia of PVAT and infiltration of proinflammatory immune cells in PVAT have been demonstrated in obesity and insulin resistance (25). Maladaptive responses of PVAT also contribute to low-grade vascular inflammation and impaired endothelial function and vascular stiffness (33, 127). PVAT loses its normal vascular protective effects in obesity and insulin resistance (53). Enhanced infiltration of inflammatory immune cells in PVAT is associated with endothelial dysfunction (44). Dysfunctional PVAT adjacent to the vessel wall promotes vascular stiffness through secretion of several vasoactive factors, including aldosterone. In this regard, all components of the RAAS, including ANG II and angiotensin-(1–7) and their receptors, are expressed in PVAT. Treatment with an angiotensin receptor antagonist significantly lowered adipose tissue weight and improved PVAT-mediated vascular relaxation (72). Moreover, administration of the MR antagonist spironolactone also prevented endothelial NO uncoupling caused by β-adrenergic stimulation in a PVAT-dependent manner (133). Increased levels of IL-6 in PVAT are seen in obesity, and aortic PVAT-derived IL-6 contributes to arterial stiffness in LDL receptor-deficient mice (29). Decreased levels of adiponectin in PVAT also contribute to impaired vascular relaxation and promotion of vascular stiffness (53, 69, 134). Recent studies have demonstrated an important role for PVAT-produced adiponectin in attenuating vascular oxidase stress and vascular stiffness (2). Diet-induced obesity is also associated with increased arginase activity, which leads to l-arginine deficiency in PVAT and uncoupling of eNOS, thereby resulting in reduced bioavailable NO (142). Decreased bioavailable NO causes activation of TG2 and arterial stiffness (5). In addition to adipokines and NO, an unidentified adipocyte-derived relaxing factor appears to play an important role in the regulation of vascular tone by PVAT. The adipocyte-derived relaxing factors released by PVAT cause vasorelaxation, which occurs by promoting the opening of voltage-dependent K+ channels in the plasma membrane of smooth muscle cells (43, 76). The vascular relaxing properties of PVAT are abrogated in obesity. Indeed, PVAT from obese rats with metabolic syndrome was observed to promote impaired mesenteric vascular relaxation mediated via prostaglandins (89). Aortic PVAT from diet-induced obese mice potentiates vascular contractility through cyclooxygenase-derived adipose tissue-derived contracting factor (90). Furthermore, PVAT from obese swine augmented KCl-induced coronary contractions (100). These obese animal PVAT-mediated abnormal vascular responses were abolished in the presence of inhibitors of Ca2+ channels. Obese PVAT also inhibits ATP-sensitive K+ channel-mediated dilation in lean and obese coronary vessels (98). Together, these studies support the idea that maladaptive adipose tissue remodeling has a critical role in reducing vascular relaxation and promoting arterial stiffness in obesity.

SEX DIFFERENCES IN CV STIFFNESS

There is emerging evidence in obese and diabetic females that development of systemic and tissue insulin resistance is a strong risk factor for CV stiffness and CVD (24, 31, 110). Females lose the CVD protection normally afforded to them when they are obese, insulin-resistant, and diabetic (26, 81). Obese women have substantially increased vascular stiffness compared with age-matched men. The mechanisms for the abrogation of CV protection in premenopausal obese and diabetic women likely involve a number of mechanisms (109). Cross talk between MRs and estrogen receptors (ERs) may promote expression and activation of vascular ENaC. Indeed, binding of 27-hydroxycholesterol to ERα is one mechanism that may contribute to deleterious effects on arterial stiffness in females with obesity and insulin resistance (82, 130, 131, 147). We have developed a translationally relevant model of WD-induced CV stiffness and diastolic dysfunction, studied insulin resistance in WD-fed females, and demonstrated abrogation of the female advantage in the setting of obesity. We have further explored the role of elevated aldosterone levels and MR activation in CV dysfunction and vascular stiffness in WD-fed females. In contrast to studies where females are protected in a high-fat-diet model, females fed a high-saturated-fat–high-refined-carbohydrate WD exhibit systemic and CV tissue insulin resistance and cardiac and vascular stiffness. We observed that a WD has an earlier adverse impact on CV tissue insulin metabolic signaling in females than males (81), and this impaired insulin signaling was associated with increased CV stiffness (26). Elevated plasma aldosterone levels were accompanied by increased CV MR expression in WD-fed female mice (26, 56). MR antagonism with very low-dose spironolactone administration prevented this diet-induced CV stiffness in females (26). Furthermore, endothelial-specific deletion of MRs prevented WD-induced aortic stiffness in concert with decreased expression of ENaC and increased bioavailable NO (56). Recently, we demonstrated an improvement in arterial stiffness in WD-fed EC ERα KO mice, suggesting that ERα signaling contributes to this arterial stiffness (82).

AGING

Two-thirds of American adults are overweight or obese (121). Obesity contributes to CVD in all age groups, including those >50 yr of age (126, 128). Arterial stiffening drives an increase in systolic pressure compared with diastolic pressure with increasing age (45). Although arterial stiffening with aging is usually attributed to increases in collagen deposition, reductions in and breaking of elastin also contribute to this process. Furthermore, increased cross-linking of connective tissue promoted by TG2 and AGEs is also important in promoting this aging process (61, 118). Enhanced oxidative stress and inflammation are seen in both age- and obesity-associated arterial stiffening (136). Enhanced PVAT generation of ROS has been reported in aging as well, thereby raising the possibility of PVAT-derived ROS as a potential mediator of arterial stiffness and the inflammatory response in aging processes accelerated with obesity (18, 134).

CONCLUSIONS

Arterial stiffness in obesity is emerging as an independent risk factor promoting the progression of CVD and CKD. Interactions of excess caloric intake with inappropriately activated RAAS and inflammation promote vascular insulin resistance, decreased bioavailable NO, maladaptive immune inflammatory response, and dysfunctional PVAT, all of which promote cardiac and vascular stiffness. High dietary fructose-induced production of uric acid and enhanced tissue xanthine oxidase activity amplify vascular inflammation and promote arterial stiffness. ECM remodeling and connective tissue cross-linking contribute to arterial stiffness in obesity. Recent studies highlight the role of cell-specific MR activation in the development of arterial stiffness in diet-induced obesity. Recent investigation in our laboratory also implies that increased signaling through the endothelial MR in association with WD consumption may also paradoxically increase EnNaC activation, thereby promoting vascular stiffness. The mechanisms revealed by these studies may help drive preventive and therapeutic strategies directed at CV stiffness that will reduce the inordinate CVD risk in overweight persons.

GRANTS

This work was supported by National Heart, Lung, and Blood Institute Grants HL-073101 and HL-107910 (to J. R. Stanton) and Department of Veterans Affairs Biomedical Laboratory Research and Development Grant 0018 (to J. R. Stanton).

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

A.R.A. and G.J. prepared figures; A.R.A., G.J., and J.R.S. drafted manuscript; A.R.A., G.J., and J.R.S. edited and revised manuscript; A.R.A., G.J., and J.R.S. approved final version of manuscript.

ACKNOWLEDGMENTS

The authors thank Brenda Hunter for editorial assistance and Brady Barron for supervising the research conducted in our laboratory.

REFERENCES

  • 1. Abraham NG, Junge JM, Drummond GS. Translational significance of heme oxygenase in obesity and metabolic syndrome. Trends Pharmacol Sci 37: 17–36, 2016. doi:10.1016/j.tips.2015.09.003.
    Crossref | PubMed | Web of Science | Google Scholar
  • 2. Antonopoulos AS, Margaritis M, Coutinho P, Shirodaria C, Psarros C, Herdman L, Sanna F, De Silva R, Petrou M, Sayeed R, Krasopoulos G, Lee R, Digby J, Reilly S, Bakogiannis C, Tousoulis D, Kessler B, Casadei B, Channon KM, Antoniades C. Adiponectin as a link between type 2 diabetes and vascular NADPH oxidase activity in the human arterial wall: the regulatory role of perivascular adipose tissue. Diabetes 64: 2207–2219, 2015. doi:10.2337/db14-1011.
    Crossref | PubMed | Web of Science | Google Scholar
  • 3. Aroor A, McKarns S, Nistala R, Demarco V, Gardner M, Garcia-Touza M, Whaley-Connell A, Sowers JR. DPP-4 inhibitors as therapeutic modulators of immune cell function and associated cardiovascular and renal insulin resistance in obesity and diabetes. Cardiorenal Med 3: 48–56, 2013. doi:10.1159/000348756.
    Crossref | PubMed | Web of Science | Google Scholar
  • 5. Aroor AR, Demarco VG, Jia G, Sun Z, Nistala R, Meininger GA, Sowers JR. The role of tissue renin-angiotensin-aldosterone system in the development of endothelial dysfunction and arterial stiffness. Front Endocrinol (Lausanne) 4: 161, 2013. doi:10.3389/fendo.2013.00161.
    Crossref | PubMed | Google Scholar
  • 6. Aroor AR, Jia G, Habibi J, Sun Z, Ramirez-Perez FI, Brady B, Chen D, Martinez-Lemus LA, Manrique C, Nistala R, Whaley-Connell AT, Demarco VG, Meininger GA, Sowers JR. Uric acid promotes vascular stiffness, maladaptive inflammatory responses and proteinuria in Western diet fed mice. Metabolism 74: 32–40, 2017. doi:10.1016/j.metabol.2017.06.006.
    Crossref | PubMed | Web of Science | Google Scholar
  • 7. Aroor AR, Mandavia C, Ren J, Sowers JR, Pulakat L. Mitochondria and oxidative stress in the cardiorenal metabolic syndrome. Cardiorenal Med 2: 87–109, 2012. doi:10.1159/000335675.
    Crossref | PubMed | Web of Science | Google Scholar
  • 8. Aroor AR, Mandavia CH, Sowers JR. Insulin resistance and heart failure: molecular mechanisms. Heart Fail Clin 8: 609–617, 2012. doi:10.1016/j.hfc.2012.06.005.
    Crossref | PubMed | Web of Science | Google Scholar
  • 9. Aroor AR, McKarns S, Demarco VG, Jia G, Sowers JR. Maladaptive immune and inflammatory pathways lead to cardiovascular insulin resistance. Metabolism 62: 1543–1552, 2013. doi:10.1016/j.metabol.2013.07.001.
    Crossref | PubMed | Web of Science | Google Scholar
  • 10. Aroor AR, Sowers JR, Jia G, DeMarco VG. Pleiotropic effects of the dipeptidylpeptidase-4 inhibitors on the cardiovascular system. Am J Physiol Heart Circ Physiol 307: H477–H492, 2014. doi:10.1152/ajpheart.00209.2014.
    Link | Web of Science | Google Scholar
  • 11. Baldo MP, Forechi L, Morra EA, Zaniqueli D, Machado RC, Lunz W, Rodrigues SL, Mill JG. Long-term use of low-dose spironolactone in spontaneously hypertensive rats: effects on left ventricular hypertrophy and stiffness. Pharmacol Rep 63: 975–982, 2011. doi:10.1016/S1734-1140(11)70613-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 12. Belo VA, Guimarães DA, Castro MM. Matrix metalloproteinase 2 as a potential mediator of vascular smooth muscle cell migration and chronic vascular remodeling in hypertension. J Vasc Res 52: 221–231, 2015. doi:10.1159/000441621.
    Crossref | PubMed | Web of Science | Google Scholar
  • 13. Belo VA, Lacchini R, Miranda JA, Lanna CM, Souza-Costa DC, Tanus-Santos JE. Increased activity of MMP-2 in hypertensive obese children is associated with hypoadiponectinemia. Obesity (Silver Spring) 23: 177–182, 2015. doi:10.1002/oby.20939.
    Crossref | PubMed | Web of Science | Google Scholar
  • 14. Bender SB, McGraw AP, Jaffe IZ, Sowers JR. Mineralocorticoid receptor-mediated vascular insulin resistance: an early contributor to diabetes-related vascular disease? Diabetes 62: 313–319, 2013. doi:10.2337/db12-0905.
    Crossref | PubMed | Web of Science | Google Scholar
  • 15. Bene NC, Alcaide P, Wortis HH, Jaffe IZ. Mineralocorticoid receptors in immune cells: emerging role in cardiovascular disease. Steroids 91: 38–45, 2014. doi:10.1016/j.steroids.2014.04.005.
    Crossref | PubMed | Web of Science | Google Scholar
  • 16. Bostick B, Habibi J, DeMarco VG, Jia G, Domeier TL, Lambert MD, Aroor AR, Nistala R, Bender SB, Garro M, Hayden MR, Ma L, Manrique C, Sowers JR. Mineralocorticoid receptor blockade prevents Western diet-induced diastolic dysfunction in female mice. Am J Physiol Heart Circ Physiol 308: H1126–H1135, 2015. doi:10.1152/ajpheart.00898.2014.
    Link | Web of Science | Google Scholar
  • 17. Brown NK, Zhou Z, Zhang J, Zeng R, Wu J, Eitzman DT, Chen YE, Chang L. Perivascular adipose tissue in vascular function and disease: a review of current research and animal models. Arterioscler Thromb Vasc Biol 34: 1621–1630, 2014. doi:10.1161/ATVBAHA.114.303029.
    Crossref | PubMed | Web of Science | Google Scholar
  • 18. Brunner EJ, Shipley MJ, Ahmadi-Abhari S, Tabak AG, McEniery CM, Wilkinson IB, Marmot MG, Singh-Manoux A, Kivimaki M. Adiposity, obesity, and arterial aging: longitudinal study of aortic stiffness in the Whitehall II cohort. Hypertension 66: 294–300, 2015. doi:10.1161/HYPERTENSIONAHA.115.05494.
    Crossref | PubMed | Web of Science | Google Scholar
  • 19. Candela J, Wang R, White C. Microvascular endothelial dysfunction in obesity is driven by macrophage-dependent hydrogen sulfide depletion. Arterioscler Thromb Vasc Biol 37: 889–899, 2017. doi:10.1161/ATVBAHA.117.309138.
    Crossref | PubMed | Web of Science | Google Scholar
  • 20. Chaudhary K, Malhotra K, Sowers J, Aroor A. Uric acid—key ingredient in the recipe for cardiorenal metabolic syndrome. Cardiorenal Med 3: 208–220, 2013. doi:10.1159/000355405.
    Crossref | PubMed | Web of Science | Google Scholar
  • 21. Cooper LL, Mitchell GF. Aortic stiffness, cerebrovascular dysfunction, and memory. Pulse (Basel) 4: 69–77, 2016. doi:10.1159/000448176.
    Crossref | PubMed | Google Scholar
  • 22. Cooper LL, Palmisano JN, Benjamin EJ, Larson MG, Vasan RS, Mitchell GF, Hamburg NM. Microvascular function contributes to the relation between aortic stiffness and cardiovascular events: the Framingham Heart Study. Circ Cardiovasc Imaging 9: e004979, 2016. doi:10.1161/CIRCIMAGING.116.004979.
    Crossref | PubMed | Web of Science | Google Scholar
  • 23. Cote AT, Phillips AA, Harris KC, Sandor GG, Panagiotopoulos C, Devlin AM. Obesity and arterial stiffness in children: systematic review and meta-analysis. Arterioscler Thromb Vasc Biol 35: 1038–1044, 2015. doi:10.1161/ATVBAHA.114.305062.
    Crossref | PubMed | Web of Science | Google Scholar
  • 24. Coutinho T, Borlaug BA, Pellikka PA, Turner ST, Kullo IJ. Sex differences in arterial stiffness and ventricular-arterial interactions. J Am Coll Cardiol 61: 96–103, 2013. doi:10.1016/j.jacc.2012.08.997.
    Crossref | PubMed | Web of Science | Google Scholar
  • 25. DeMarco VG, Aroor AR, Sowers JR. The pathophysiology of hypertension in patients with obesity. Nat Rev Endocrinol 10: 364–376, 2014. doi:10.1038/nrendo.2014.44.
    Crossref | PubMed | Web of Science | Google Scholar
  • 26. DeMarco VG, Habibi J, Jia G, Aroor AR, Ramirez-Perez FI, Martinez-Lemus LA, Bender SB, Garro M, Hayden MR, Sun Z, Meininger GA, Manrique C, Whaley-Connell A, Sowers JR. Low-dose mineralocorticoid receptor blockade prevents Western diet-induced arterial stiffening in female mice. Hypertension 66: 99–107, 2015. doi:10.1161/HYPERTENSIONAHA.115.05674.
    Crossref | PubMed | Web of Science | Google Scholar
  • 27. Deng G, Qiu Z, Li D, Fang Y, Zhang S. Effects of allopurinol on arterial stiffness: a meta-analysis of randomized controlled trials. Med Sci Monit 22: 1389–1397, 2016. doi:10.12659/MSM.898370.
    Crossref | PubMed | Web of Science | Google Scholar
  • 28. Drüppel V, Kusche-Vihrog K, Grossmann C, Gekle M, Kasprzak B, Brand E, Pavenstädt H, Oberleithner H, Kliche K. Long-term application of the aldosterone antagonist spironolactone prevents stiff endothelial cell syndrome. FASEB J 27: 3652–3659, 2013. doi:10.1096/fj.13-228312.
    Crossref | PubMed | Web of Science | Google Scholar
  • 29. Du B, Ouyang A, Eng JS, Fleenor BS. Aortic perivascular adipose-derived interleukin-6 contributes to arterial stiffness in low-density lipoprotein receptor deficient mice. Am J Physiol Heart Circ Physiol 308: H1382–H1390, 2015. doi:10.1152/ajpheart.00712.2014.
    Link | Web of Science | Google Scholar
  • 30. Dugani S, Gaziano TA. 25 by 25: Achieving global reduction in cardiovascular mortality. Curr Cardiol Rep 18: 10, 2016. doi:10.1007/s11886-015-0679-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 31. Duprez D, Jacobs DR Jr. Arterial stiffness and left ventricular diastolic function: does sex matter? Hypertension 60: 283–284, 2012. doi:10.1161/HYPERTENSIONAHA.112.197616.
    Crossref | PubMed | Web of Science | Google Scholar
  • 32. Durante W. Protective role of heme oxygenase-1 against inflammation in atherosclerosis. Front Biosci 16: 2372–2388, 2011. doi:10.2741/3860.
    Crossref | PubMed | Web of Science | Google Scholar
  • 33. Eringa EC, Bakker W, van Hinsbergh VW. Paracrine regulation of vascular tone, inflammation and insulin sensitivity by perivascular adipose tissue. Vascul Pharmacol 56: 204–209, 2012. doi:10.1016/j.vph.2012.02.003.
    Crossref | PubMed | Web of Science | Google Scholar
  • 34. Fels J, Jeggle P, Liashkovich I, Peters W, Oberleithner H. Nanomechanics of vascular endothelium. Cell Tissue Res 355: 727–737, 2014. doi:10.1007/s00441-014-1853-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 35. Femia R, Kozakova M, Nannipieri M, Gonzales-Villalpando C, Stern MP, Haffner SM, Ferrannini E. Carotid intima-media thickness in confirmed prehypertensive subjects: predictors and progression. Arterioscler Thromb Vasc Biol 27: 2244–2249, 2007. doi:10.1161/ATVBAHA.107.149641.
    Crossref | PubMed | Web of Science | Google Scholar
  • 36. Fernhall B, Agiovlasitis S. Arterial function in youth: window into cardiovascular risk. J Appl Physiol (1985) 105: 325–333, 2008. doi:10.1152/japplphysiol.00001.2008.
    Link | Web of Science | Google Scholar
  • 37. Fleenor BS, Eng JS, Sindler AL, Pham BT, Kloor JD, Seals DR. Superoxide signaling in perivascular adipose tissue promotes age-related artery stiffness. Aging Cell 13: 576–578, 2014. doi:10.1111/acel.12196.
    Crossref | PubMed | Web of Science | Google Scholar
  • 38. Foote CA, Castorena-Gonzalez JA, Staiculescu MC, Clifford PS, Hill MA, Meininger GA, Martinez-Lemus LA. Brief serotonin exposure initiates arteriolar inward remodeling processes in vivo that involve transglutaminase activation and actin cytoskeleton reorganization. Am J Physiol Heart Circ Physiol 310: H188–H198, 2016. doi:10.1152/ajpheart.00666.2015.
    Link | Web of Science | Google Scholar
  • 39. Ford ML, Tomlinson LA, Chapman TP, Rajkumar C, Holt SG. Aortic stiffness is independently associated with rate of renal function decline in chronic kidney disease stages 3 and 4. Hypertension 55: 1110–1115, 2010. doi:10.1161/HYPERTENSIONAHA.109.143024.
    Crossref | PubMed | Web of Science | Google Scholar
  • 40. Galmiche G, Pizard A, Gueret A, El Moghrabi S, Ouvrard-Pascaud A, Berger S, Challande P, Jaffe IZ, Labat C, Lacolley P, Jaisser F. Smooth muscle cell mineralocorticoid receptors are mandatory for aldosterone-salt to induce vascular stiffness. Hypertension 63: 520–526, 2014. doi:10.1161/HYPERTENSIONAHA.113.01967.
    Crossref | PubMed | Web of Science | Google Scholar
  • 41. Gálvez B, de Castro J, Herold D, Dubrovska G, Arribas S, González MC, Aranguez I, Luft FC, Ramos MP, Gollasch M, Fernández Alfonso MS. Perivascular adipose tissue and mesenteric vascular function in spontaneously hypertensive rats. Arterioscler Thromb Vasc Biol 26: 1297–1302, 2006. doi:10.1161/01.ATV.0000220381.40739.dd.
    Crossref | PubMed | Web of Science | Google Scholar
  • 42. Ghanim H, Aljada A, Hofmeyer D, Syed T, Mohanty P, Dandona P. Circulating mononuclear cells in the obese are in a proinflammatory state. Circulation 110: 1564–1571, 2004. doi:10.1161/01.CIR.0000142055.53122.FA.
    Crossref | PubMed | Web of Science | Google Scholar
  • 43. Gollasch M. Vasodilator signals from perivascular adipose tissue. Br J Pharmacol 165: 633–642, 2012. doi:10.1111/j.1476-5381.2011.01430.x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 44. Gómez-Hernández A, Beneit N, Díaz-Castroverde S, Escribano Ó. Differential role of adipose tissues in obesity and related metabolic and vascular complications. Int J Endocrinol 2016: 1216783, 2016. doi:10.1155/2016/1216783.
    Crossref | PubMed | Web of Science | Google Scholar
  • 45. Harvey A, Montezano AC, Lopes RA, Rios F, Touyz RM. Vascular fibrosis in aging and hypertension: molecular mechanisms and clinical implications. Can J Cardiol 32: 659–668, 2016. doi:10.1016/j.cjca.2016.02.070.
    Crossref | PubMed | Web of Science | Google Scholar
  • 46. Henao-Mejia J, Elinav E, Thaiss CA, Flavell RA. Inflammasomes and metabolic disease. Annu Rev Physiol 76: 57–78, 2014. doi:10.1146/annurev-physiol-021113-170324.
    Crossref | PubMed | Web of Science | Google Scholar
  • 47. Hong Z, Sun Z, Li Z, Mesquitta WT, Trzeciakowski JP, Meininger GA. Coordination of fibronectin adhesion with contraction and relaxation in microvascular smooth muscle. Cardiovasc Res 96: 73–80, 2012. doi:10.1093/cvr/cvs239.
    Crossref | PubMed | Web of Science | Google Scholar
  • 48. Hosick PA, Stec DE. Heme oxygenase, a novel target for the treatment of hypertension and obesity? Am J Physiol Regul Integr Comp Physiol 302: R207–R214, 2012. doi:10.1152/ajpregu.00517.2011.
    Link | Web of Science | Google Scholar
  • 49. Hua Y, Nair S. Proteases in cardiometabolic diseases: pathophysiology, molecular mechanisms and clinical applications. Biochim Biophys Acta 1852: 195–208, 2015. doi:10.1016/j.bbadis.2014.04.032.
    Crossref | PubMed | Web of Science | Google Scholar
  • 50. Idriss NK, Blann AD, Lip GY. Hemoxygenase-1 in cardiovascular disease. J Am Coll Cardiol 52: 971–978, 2008. doi:10.1016/j.jacc.2008.06.019.
    Crossref | PubMed | Web of Science | Google Scholar
  • 51. Jaffe IZ, Jaisser F. Endothelial cell mineralocorticoid receptors: turning cardiovascular risk factors into cardiovascular dysfunction. Hypertension 63: 915–917, 2014. doi:10.1161/HYPERTENSIONAHA.114.01997.
    Crossref | PubMed | Web of Science | Google Scholar
  • 52. Jeggle P, Callies C, Tarjus A, Fassot C, Fels J, Oberleithner H, Jaisser F, Kusche-Vihrog K. Epithelial sodium channel stiffens the vascular endothelium in vitro and in Liddle mice. Hypertension 61: 1053–1059, 2013. doi:10.1161/HYPERTENSIONAHA.111.199455.
    Crossref | PubMed | Web of Science | Google Scholar
  • 53. Jia G, Aroor AR, DeMarco VG, Martinez-Lemus LA, Meininger GA, Sowers JR. Vascular stiffness in insulin resistance and obesity. Front Physiol 6: 231, 2015. doi:10.3389/fphys.2015.00231.
    Crossref | PubMed | Web of Science | Google Scholar
  • 54. Jia G, Aroor AR, Sowers JR. Arterial stiffness: a nexus between cardiac and renal disease. Cardiorenal Med 4: 60–71, 2014. doi:10.1159/000360867.
    Crossref | PubMed | Web of Science | Google Scholar
  • 55. Jia G, Aroor AR, Whaley-Connell AT, Sowers JR. Fructose and uric acid: is there a role in endothelial function? Curr Hypertens Rep 16: 434, 2014. doi:10.1007/s11906-014-0434-z.
    Crossref | PubMed | Web of Science | Google Scholar
  • 56. Jia G, Habibi J, Aroor AR, Martinez-Lemus LA, DeMarco VG, Ramirez-Perez FI, Sun Z, Hayden MR, Meininger GA, Mueller KB, Jaffe IZ, Sowers JR. Endothelial mineralocorticoid receptor mediates diet-induced aortic stiffness in females. Circ Res 118: 935–943, 2016. doi:10.1161/CIRCRESAHA.115.308269.
    Crossref | PubMed | Web of Science | Google Scholar
  • 57. Jia G, Habibi J, Bostick BP, Ma L, DeMarco VG, Aroor AR, Hayden MR, Whaley-Connell AT, Sowers JR. Uric acid promotes left ventricular diastolic dysfunction in mice fed a Western diet. Hypertension 65: 531–539, 2015. doi:10.1161/HYPERTENSIONAHA.114.04737.
    Crossref | PubMed | Web of Science | Google Scholar
  • 58. Jia G, Habibi J, DeMarco VG, Martinez-Lemus LA, Ma L, Whaley-Connell AT, Aroor AR, Domeier TL, Zhu Y, Meininger GA, Mueller KB, Jaffe IZ, Sowers JR. Endothelial mineralocorticoid receptor deletion prevents diet-induced cardiac diastolic dysfunction in females. Hypertension 66: 1159–1167, 2015. doi:10.1161/HYPERTENSIONAHA.115.06015.
    Crossref | PubMed | Web of Science | Google Scholar
  • 59. Jia G, Jia Y, Sowers JR. Contribution of maladaptive adipose tissue expansion to development of cardiovascular disease. Compr Physiol 7: 253–262, 2016. doi:10.1002/cphy.c160014.
    Crossref | PubMed | Web of Science | Google Scholar
  • 60. Johnson BD, Mather KJ, Wallace JP. Mechanotransduction of shear in the endothelium: basic studies and clinical implications. Vasc Med 16: 365–377, 2011. doi:10.1177/1358863X11422109.
    Crossref | PubMed | Web of Science | Google Scholar
  • 61. Jung SM, Jandu S, Steppan J, Belkin A, An SS, Pak A, Choi EY, Nyhan D, Butlin M, Viegas K, Avolio A, Berkowitz DE, Santhanam L. Increased tissue transglutaminase activity contributes to central vascular stiffness in eNOS knockout mice. Am J Physiol Heart Circ Physiol 305: H803–H810, 2013. doi:10.1152/ajpheart.00103.2013.
    Link | Web of Science | Google Scholar
  • 62. Kalupahana NS, Moustaid-Moussa N, Claycombe KJ. Immunity as a link between obesity and insulin resistance. Mol Aspects Med 33: 26–34, 2012. doi:10.1016/j.mam.2011.10.011.
    Crossref | PubMed | Web of Science | Google Scholar
  • 63. Kassan M, Galan M, Partyka M, Trebak M, Matrougui K. Interleukin-10 released by CD4+CD25+ natural regulatory T cells improves microvascular endothelial function through inhibition of NADPH oxidase activity in hypertensive mice. Arterioscler Thromb Vasc Biol 31: 2534–2542, 2011. doi:10.1161/ATVBAHA.111.233262.
    Crossref | PubMed | Web of Science | Google Scholar
  • 64. Kellow NJ, Coughlan MT. Effect of diet-derived advanced glycation end products on inflammation. Nutr Rev 73: 737–759, 2015. doi:10.1093/nutrit/nuv030.
    Crossref | PubMed | Web of Science | Google Scholar
  • 65. Kim JA, Jang HJ, Martinez-Lemus LA, Sowers JR. Activation of mTOR/p70S6 kinase by ANG II inhibits insulin-stimulated endothelial nitric oxide synthase and vasodilation. Am J Physiol Endocrinol Metab 302: E201–E208, 2012. doi:10.1152/ajpendo.00497.2011.
    Link | Web of Science | Google Scholar
  • 66. Kithas PA, Supiano MA. Spironolactone and hydrochlorothiazide decrease vascular stiffness and blood pressure in geriatric hypertension. J Am Geriatr Soc 58: 1327–1332, 2010. doi:10.1111/j.1532-5415.2010.02905.x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 67. Kumar R, Thomas CM, Yong QC, Chen W, Baker KM. The intracrine renin-angiotensin system. Clin Sci (Lond) 123: 273–284, 2012. doi:10.1042/CS20120089.
    Crossref | PubMed | Web of Science | Google Scholar
  • 68. Lang F. Stiff endothelial cell syndrome in vascular inflammation and mineralocorticoid excess. Hypertension 57: 146–147, 2011. doi:10.1161/HYPERTENSIONAHA.110.164558.
    Crossref | PubMed | Web of Science | Google Scholar
  • 69. Lastra G, Manrique C. Perivascular adipose tissue, inflammation and insulin resistance: link to vascular dysfunction and cardiovascular disease. Horm Mol Biol Clin Investig 22: 19–26, 2015. doi:10.1515/hmbci-2015-0010.
    Crossref | PubMed | Web of Science | Google Scholar
  • 70. Lastra G, Manrique C, Jia G, Aroor AR, Hayden MR, Barron BJ, Niles B, Padilla J, Sowers JR. Xanthine oxidase inhibition protects against Western diet-induced aortic stiffness and impaired vasorelaxation in female mice. Am J Physiol Regul Integr Comp Physiol 313: R67–R77, 2017. doi:10.1152/ajpregu.00483.2016.
    Link | Web of Science | Google Scholar
  • 71. Lee WJ, Tateya S, Cheng AM, Rizzo-DeLeon N, Wang NF, Handa P, Wilson CL, Clowes AW, Sweet IR, Bomsztyk K, Schwartz MW, Kim F. M2 macrophage polarization mediates anti-inflammatory effects of endothelial nitric oxide signaling. Diabetes 64: 2836–2846, 2015. doi:10.2337/db14-1668.
    Crossref | PubMed | Web of Science | Google Scholar
  • 72. Li H, Li M, Liu P, Wang Y, Zhang H, Li H, Yang S, Song Y, Yin Y, Gao L, Cheng S, Cai J, Tian G. Telmisartan ameliorates nephropathy in metabolic syndrome by reducing leptin release from perirenal adipose tissue. Hypertension 68: 478–490, 2016. doi:10.1161/HYPERTENSIONAHA.116.07008.
    Crossref | PubMed | Web of Science | Google Scholar
  • 73. Li R, Zhang H, Wang W, Wang X, Huang Y, Huang C, Gao F. Vascular insulin resistance in prehypertensive rats: role of PI3-kinase/Akt/eNOS signaling. Eur J Pharmacol 628: 140–147, 2010. doi:10.1016/j.ejphar.2009.11.038.
    Crossref | PubMed | Web of Science | Google Scholar
  • 74. Liao J, Farmer J. Arterial stiffness as a risk factor for coronary artery disease. Curr Atheroscler Rep 16: 387, 2014. doi:10.1007/s11883-013-0387-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 75. Liu SL, Bae YH, Yu C, Monslow J, Hawthorne EA, Castagnino P, Branchetti E, Ferrari G, Damrauer SM, Puré E, Assoian RK. Matrix metalloproteinase-12 is an essential mediator of acute and chronic arterial stiffening. Sci Rep 5: 17189, 2015. doi:10.1038/srep17189.
    Crossref | PubMed | Web of Science | Google Scholar
  • 76. Löhn M, Dubrovska G, Lauterbach B, Luft FC, Gollasch M, Sharma AM. Periadventitial fat releases a vascular relaxing factor. FASEB J 16: 1057–1063, 2002. doi:10.1096/fj.02-0024com.
    Crossref | PubMed | Web of Science | Google Scholar
  • 77. Loufrani L, Henrion D. Role of the cytoskeleton in flow (shear stress)-induced dilation and remodeling in resistance arteries. Med Biol Eng Comput 46: 451–460, 2008. doi:10.1007/s11517-008-0306-2.
    Crossref | PubMed | Web of Science | Google Scholar
  • 78. Luft FC. Molecular mechanisms of arterial stiffness: new insights. J Am Soc Hypertens 6: 436–438, 2012. doi:10.1016/j.jash.2012.10.004.
    Crossref | PubMed | Web of Science | Google Scholar
  • 79. Magenta A, Greco S, Capogrossi MC, Gaetano C, Martelli F. Nitric oxide, oxidative stress, and p66Shc interplay in diabetic endothelial dysfunction. BioMed Res Int 2014: 193095, 2014. doi:10.1155/2014/193095.
    Crossref | PubMed | Web of Science | Google Scholar
  • 80. Mancia G, Fagard R. Guidelines for the management of hypertension and target organ damage: reply [reply]. J Hypertens 31: 2464–2465, 2013. doi:10.1097/HJH.0000000000000006.
    Crossref | PubMed | Web of Science | Google Scholar
  • 81. Manrique C, DeMarco VG, Aroor AR, Mugerfeld I, Garro M, Habibi J, Hayden MR, Sowers JR. Obesity and insulin resistance induce early development of diastolic dysfunction in young female mice fed a Western diet. Endocrinology 154: 3632–3642, 2013. doi:10.1210/en.2013-1256.
    Crossref | PubMed | Web of Science | Google Scholar
  • 82. Manrique C, Lastra G, Ramirez-Perez FI, Haertling D, DeMarco VG, Aroor AR, Jia G, Chen D, Barron BJ, Garro M, Padilla J, Martinez-Lemus LA, Sowers JR. Endothelial estrogen receptor-α does not protect against vascular stiffness induced by Western diet in female mice. Endocrinology 157: 1590–1600, 2016. doi:10.1210/en.2015-1681.
    Crossref | PubMed | Web of Science | Google Scholar
  • 83. Marti CN, Gheorghiade M, Kalogeropoulos AP, Georgiopoulou VV, Quyyumi AA, Butler J. Endothelial dysfunction, arterial stiffness, and heart failure. J Am Coll Cardiol 60: 1455–1469, 2012. doi:10.1016/j.jacc.2011.11.082.
    Crossref | PubMed | Web of Science | Google Scholar
  • 84. Martinez-Lemus LA, Aroor AR, Ramirez-Perez FI, Jia G, Habibi J, DeMarco VG, Barron B, Whaley-Connell A, Nistala R, Sowers JR. Amiloride improves endothelial function and reduces vascular stiffness in female mice fed a Western diet. Front Physiol 8: 456, 2017. doi:10.3389/fphys.2017.00456.
    Crossref | PubMed | Web of Science | Google Scholar
  • 85. Martínez-Revelles S, García-Redondo AB, Avendaño MS, Varona S, Palao T, Orriols M, Roque FR, Fortuño A, Touyz RM, Martínez-González J, Salaices M, Rodríguez C, Briones AM. Lysyl oxidase induces vascular oxidative stress and contributes to arterial stiffness and abnormal elastin structure in hypertension: role of p38MAPK. Antioxid Redox Signal 27: 379–397, 2017. doi:10.1089/ars.2016.6642.
    Crossref | PubMed | Web of Science | Google Scholar
  • 86. Matsuoka O, Otsuka K, Murakami S, Hotta N, Yamanaka G, Kubo Y, Yamanaka T, Shinagawa M, Nunoda S, Nishimura Y, Shibata K, Saitoh H, Nishinaga M, Ishine M, Wada T, Okumiya K, Matsubayashi K, Yano S, Ichihara K, Cornélissen G, Halberg F, Ozawa T. Arterial stiffness independently predicts cardiovascular events in an elderly community. Longitudinal Investigation for the Longevity and Aging in Hokkaido County (LILAC) Study. Biomed Pharmacother 59 Suppl 1: S40–S44, 2005. doi:10.1016/S0753-3322(05)80008-3.
    Crossref | PubMed | Web of Science | Google Scholar
  • 87. McCullough AJ. Epidemiology of the metabolic syndrome in the USA. J Dig Dis 12: 333–340, 2011. doi:10.1111/j.1751-2980.2010.00469.x.
    Crossref | PubMed | Web of Science | Google Scholar
  • 88. McCurley A, McGraw A, Pruthi D, Jaffe IZ. Smooth muscle cell mineralocorticoid receptors: role in vascular function and contribution to cardiovascular disease. Pflugers Arch 465: 1661–1670, 2013. doi:10.1007/s00424-013-1282-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 89. Mendizábal Y, Llorens S, Nava E. Vasoactive effects of prostaglandins from the perivascular fat of mesenteric resistance arteries in WKY and SHROB rats. Life Sci 93: 1023–1032, 2013. doi:10.1016/j.lfs.2013.10.021.
    Crossref | PubMed | Web of Science | Google Scholar
  • 90. Meyer MR, Fredette NC, Barton M, Prossnitz ER. Regulation of vascular smooth muscle tone by adipose-derived contracting factor. PLoS One 8: e79245, 2013. doi:10.1371/journal.pone.0079245.
    Crossref | PubMed | Web of Science | Google Scholar
  • 91. Mitchell GF. Arterial stiffness and hypertension. Hypertension 64: 13–18, 2014. doi:10.1161/HYPERTENSIONAHA.114.00921.
    Crossref | PubMed | Web of Science | Google Scholar
  • 92. Mitchell GF. Arterial stiffness and hypertension: chicken or egg? Hypertension 64: 210–214, 2014. doi:10.1161/HYPERTENSIONAHA.114.03449.
    Crossref | PubMed | Web of Science | Google Scholar
  • 93. Muniyappa R, Sowers JR. Role of insulin resistance in endothelial dysfunction. Rev Endocr Metab Disord 14: 5–12, 2013. doi:10.1007/s11154-012-9229-1.
    Crossref | PubMed | Web of Science | Google Scholar
  • 94. Muniyappa R, Yavuz S. Metabolic actions of angiotensin II and insulin: a microvascular endothelial balancing act. Mol Cell Endocrinol 378: 59–69, 2013. doi:10.1016/j.mce.2012.05.017.
    Crossref | PubMed | Web of Science | Google Scholar
  • 95. Nar G, Soylu K, Akcay M, Gulel O, Yuksel S, Meric M, Zengin H, Erbay A, Nar R, Demircan S, Sahin M. Evaluation of the relationship between arterial blood pressure, aortic stiffness and serum endothelin-1 levels in patients with essential hypertension. Clin Exp Hypertens 35: 589–594, 2013. doi:10.3109/10641963.2013.776565.
    Crossref | PubMed | Web of Science | Google Scholar
  • 96. Nava E, Llorens S. The paracrine control of vascular motion. A historical perspective. Pharmacol Res 113: 125–145, 2016. doi:10.1016/j.phrs.2016.08.003.
    Crossref | PubMed | Web of Science | Google Scholar
  • 97. Nenna A, Spadaccio C, Lusini M, Ulianich L, Chello M, Nappi F. Basic and clinical research against advanced glycation end products (AGEs): new compounds to tackle cardiovascular disease and diabetic complications. Recent Adv Cardiovasc Drug Discov 10: 10–33, 2015. doi:10.2174/1574890110666151104120039.
    Crossref | PubMed | Google Scholar
  • 98. Noblet JN, Owen MK, Goodwill AG, Sassoon DJ, Tune JD. Lean and obese coronary perivascular adipose tissue impairs vasodilation via differential inhibition of vascular smooth muscle K+ channels. Arterioscler Thromb Vasc Biol 35: 1393–1400, 2015. doi:10.1161/ATVBAHA.115.305500.
    Crossref | PubMed | Web of Science | Google Scholar
  • 99. Nosalski R, Guzik TJ. Perivascular adipose tissue inflammation in vascular disease. Br J Pharmacol 174: 3496–3513, 2017. doi:10.1111/bph.13705.
    Crossref | PubMed | Web of Science | Google Scholar
  • 100. Owen MK, Witzmann FA, McKenney ML, Lai X, Berwick ZC, Moberly SP, Alloosh M, Sturek M, Tune JD. Perivascular adipose tissue potentiates contraction of coronary vascular smooth muscle: influence of obesity. Circulation 128: 9–18, 2013. doi:10.1161/CIRCULATIONAHA.112.001238.
    Crossref | PubMed | Web of Science | Google Scholar
  • 101. Padilla J, Vieira-Potter VJ, Jia G, Sowers JR. Role of perivascular adipose tissue on vascular reactive oxygen species in type 2 diabetes: a give-and-take relationship. Diabetes 64: 1904–1906, 2015. doi:10.2337/db15-0096.
    Crossref | PubMed | Web of Science | Google Scholar
  • 102. Pérez FR, Venegas F, González M, Andrés S, Vallejos C, Riquelme G, Sierralta J, Michea L. Endothelial epithelial sodium channel inhibition activates endothelial nitric oxide synthase via phosphoinositide 3-kinase/Akt in small-diameter mesenteric arteries. Hypertension 53: 1000–1007, 2009. doi:10.1161/HYPERTENSIONAHA.108.128520.
    Crossref | PubMed | Web of Science | Google Scholar
  • 103. Pojoga LH, Baudrand R, Adler GK. Mineralocorticoid receptor throughout the vessel: a key to vascular dysfunction in obesity. Eur Heart J 34: 3475–3477, 2013. doi:10.1093/eurheartj/eht158.
    Crossref | PubMed | Web of Science | Google Scholar
  • 104. Puddu P, Puddu GM, Cravero E, Vizioli L, Muscari A. Relationships among hyperuricemia, endothelial dysfunction and cardiovascular disease: molecular mechanisms and clinical implications. J Cardiol 59: 235–242, 2012. doi:10.1016/j.jjcc.2012.01.013.
    Crossref | PubMed | Web of Science | Google Scholar
  • 105. Rao RM, Yang L, Garcia-Cardena G, Luscinskas FW. Endothelial-dependent mechanisms of leukocyte recruitment to the vascular wall. Circ Res 101: 234–247, 2007. doi:10.1161/CIRCRESAHA.107.151860b.
    Crossref | PubMed | Web of Science | Google Scholar
  • 106. Rathinam VA, Vanaja SK, Fitzgerald KA. Regulation of inflammasome signaling. Nat Immunol 13: 333–342, 2012. doi:10.1038/ni.2237.
    Crossref | PubMed | Web of Science | Google Scholar
  • 107. Rautureau Y, Paradis P, Schiffrin EL. Cross-talk between aldosterone and angiotensin signaling in vascular smooth muscle cells. Steroids 76: 834–839, 2011. doi:10.1016/j.steroids.2011.02.015.
    Crossref | PubMed | Web of Science | Google Scholar
  • 108. Ray S, Miglio C, Eden T, Del Rio D. Assessment of vascular and endothelial dysfunction in nutritional studies. Nutr Metab Cardiovasc Dis 24: 940–946, 2014. doi:10.1016/j.numecd.2014.03.011.
    Crossref | PubMed | Web of Science | Google Scholar
  • 109. Ricchiuti V, Lian CG, Oestreicher EM, Tran L, Stone JR, Yao T, Seely EW, Williams GH, Adler GK. Estradiol increases angiotensin II type 1 receptor in hearts of ovariectomized rats. J Endocrinol 200: 75–84, 2009. doi:10.1677/JOE-08-0199.
    Crossref | PubMed | Web of Science | Google Scholar
  • 110. Russo C, Jin Z, Palmieri V, Homma S, Rundek T, Elkind MS, Sacco RL, Di Tullio MR. Arterial stiffness and wave reflection: sex differences and relationship with left ventricular diastolic function. Hypertension 60: 362–368, 2012. doi:10.1161/HYPERTENSIONAHA.112.191148.
    Crossref | PubMed | Web of Science | Google Scholar
  • 110a. Sazonova OV, Lee KL, Isenberg BC, Rich CB, Nugent MA, Wong JY. Cell-cell interactions mediate the response of vascular smooth muscle cells to substrate stiffness. Biophys J 101: 622–630, 2011. doi:10.1016/j.bpj.2011.06.051.
    Crossref | PubMed | Web of Science | Google Scholar
  • 111. Schaefer A, Hordijk PL. Cell-stiffness-induced mechanosignaling—a key driver of leukocyte transendothelial migration. J Cell Sci 128: 2221–2230, 2015. doi:10.1242/jcs.163055.
    Crossref | PubMed | Web of Science | Google Scholar
  • 112. Schiffrin EL. Immune mechanisms in hypertension and vascular injury. Clin Sci (Lond) 126: 267–274, 2014. doi:10.1042/CS20130407.
    Crossref | PubMed | Web of Science | Google Scholar
  • 113. Schiffrin EL. Mechanisms of remodelling of small arteries, antihypertensive therapy and the immune system in hypertension. Clin Invest Med 38: E394–E402, 2015. doi:10.25011/cim.v38i6.26202.
    Crossref | PubMed | Web of Science | Google Scholar
  • 114. Sehgel NL, Sun Z, Hong Z, Hunter WC, Hill MA, Vatner DE, Vatner SF, Meininger GA. Augmented vascular smooth muscle cell stiffness and adhesion when hypertension is superimposed on aging. Hypertension 65: 370–377, 2015. doi:10.1161/HYPERTENSIONAHA.114.04456.
    Crossref | PubMed | Web of Science | Google Scholar
  • 115. Sehgel NL, Vatner SF, Meininger GA. “Smooth muscle cell stiffness syndrome”—revisiting the structural basis of arterial stiffness. Front Physiol 6: 335, 2015. doi:10.3389/fphys.2015.00335.
    Crossref | PubMed | Web of Science | Google Scholar
  • 116. Sehgel NL, Zhu Y, Sun Z, Trzeciakowski JP, Hong Z, Hunter WC, Vatner DE, Meininger GA, Vatner SF. Increased vascular smooth muscle cell stiffness: a novel mechanism for aortic stiffness in hypertension. Am J Physiol Heart Circ Physiol 305: H1281–H1287, 2013. doi:10.1152/ajpheart.00232.2013.
    Link | Web of Science | Google Scholar
  • 117. Seifalian AM, Filippatos TD, Joshi J, Mikhailidis DP. Obesity and arterial compliance alterations. Curr Vasc Pharmacol 8: 155–168, 2010. doi:10.2174/157016110790886956.
    Crossref | PubMed | Web of Science | Google Scholar
  • 118. Semba RD, Nicklett EJ, Ferrucci L. Does accumulation of advanced glycation end products contribute to the aging phenotype? J Gerontol A Biol Sci Med Sci 65: 963–975, 2010. doi:10.1093/gerona/glq074.
    Crossref | PubMed | Web of Science | Google Scholar
  • 119. Shirwany NA, Zou MH. Arterial stiffness: a brief review. Acta Pharmacol Sin 31: 1267–1276, 2010. doi:10.1038/aps.2010.123.
    Crossref | PubMed | Web of Science | Google Scholar
  • 120. Sodhi K, Hilgefort J, Banks G, Gilliam C, Stevens S, Ansinelli HA, Getty M, Abraham NG, Shapiro JI, Khitan Z. Uric acid-induced adipocyte dysfunction is attenuated by HO-1 upregulation: potential role of antioxidant therapy to target obesity. Stem Cells Int 2016: 8197325, 2016. doi:10.1155/2016/8197325.
    Crossref | PubMed | Web of Science | Google Scholar
  • 121. Sowers JR. Diabetes mellitus and vascular disease. Hypertension 61: 943–947, 2013. doi:10.1161/HYPERTENSIONAHA.111.00612.
    Crossref | PubMed | Web of Science | Google Scholar
  • 122. Sowers JR, Whaley-Connell A, Hayden MR. The role of overweight and obesity in the cardiorenal syndrome. Cardiorenal Med 1: 5–12, 2011. doi:10.1159/000322822.
    Crossref | PubMed | Web of Science | Google Scholar
  • 123. Stenmark KR, Yeager ME, El Kasmi KC, Nozik-Grayck E, Gerasimovskaya EV, Li M, Riddle SR, Frid MG. The adventitia: essential regulator of vascular wall structure and function. Annu Rev Physiol 75: 23–47, 2013. doi:10.1146/annurev-physiol-030212-183802.
    Crossref | PubMed | Web of Science | Google Scholar
  • 124. Stirban A, Gawlowski T, Roden M. Vascular effects of advanced glycation endproducts: clinical effects and molecular mechanisms. Mol Metab 3: 94–108, 2013. doi:10.1016/j.molmet.2013.11.006.
    Crossref | PubMed | Web of Science | Google Scholar
  • 125. Sun Z. Aging, arterial stiffness, and hypertension. Hypertension 65: 252–256, 2015. doi:10.1161/HYPERTENSIONAHA.114.03617.
    Crossref | PubMed | Web of Science | Google Scholar
  • 126. Sutton-Tyrrell K, Newman A, Simonsick EM, Havlik R, Pahor M, Lakatta E, Spurgeon H, Vaitkevicius P. Aortic stiffness is associated with visceral adiposity in older adults enrolled in the study of health, aging, and body composition. Hypertension 38: 429–433, 2001. doi:10.1161/01.HYP.38.3.429.
    Crossref | PubMed | Web of Science | Google Scholar
  • 127. Szasz T, Bomfim GF, Webb RC. The influence of perivascular adipose tissue on vascular homeostasis. Vasc Health Risk Manag 9: 105–116, 2013. doi:10.2147/VHRM.S33760.
    Crossref | PubMed | Google Scholar
  • 128. Thorin-Trescases N, Thorin E. Lifelong cyclic mechanical strain promotes large elastic artery stiffening: increased pulse pressure and old age-related organ failure. Can J Cardiol 32: 624–633, 2016. doi:10.1016/j.cjca.2015.12.022.
    Crossref | PubMed | Web of Science | Google Scholar
  • 129. Townsend RR, Wilkinson IB, Schiffrin EL, Avolio AP, Chirinos JA, Cockcroft JR, Heffernan KS, Lakatta EG, McEniery CM, Mitchell GF, Najjar SS, Nichols WW, Urbina EM, Weber T; American Heart Association Council on Hypertension. Recommendations for improving and standardizing vascular research on arterial stiffness: a scientific statement from the American Heart Association. Hypertension 66: 698–722, 2015. doi:10.1161/HYP.0000000000000033.
    Crossref | PubMed | Web of Science | Google Scholar
  • 130. Umetani M, Domoto H, Gormley AK, Yuhanna IS, Cummins CL, Javitt NB, Korach KS, Shaul PW, Mangelsdorf DJ. 27-Hydroxycholesterol is an endogenous SERM that inhibits the cardiovascular effects of estrogen. Nat Med 13: 1185–1192, 2007. doi:10.1038/nm1641.
    Crossref | PubMed | Web of Science | Google Scholar
  • 131. Umetani M, Ghosh P, Ishikawa T, Umetani J, Ahmed M, Mineo C, Shaul PW. The cholesterol metabolite 27-hydroxycholesterol promotes atherosclerosis via proinflammatory processes mediated by estrogen receptor-α. Cell Metab 20: 172–182, 2014. doi:10.1016/j.cmet.2014.05.013.
    Crossref | PubMed | Web of Science | Google Scholar
  • 132. van den Akker J, VanBavel E, van Geel R, Matlung HL, Guvenc Tuna B, Janssen GM, van Veelen PA, Boelens WC, De Mey JG, Bakker EN. The redox state of transglutaminase 2 controls arterial remodeling. PLoS One 6: e23067, 2011. doi:10.1371/journal.pone.0023067.
    Crossref | PubMed | Web of Science | Google Scholar
  • 133. Victorio JA, Clerici SP, Palacios R, Alonso MJ, Vassallo DV, Jaffe IZ, Rossoni LV, Davel AP. Spironolactone prevents endothelial nitric oxide synthase uncoupling and vascular dysfunction induced by β-adrenergic overstimulation: role of perivascular adipose tissue. Hypertension 68: 726–735, 2016. doi:10.1161/HYPERTENSIONAHA.116.07911.
    Crossref | PubMed | Web of Science | Google Scholar
  • 134. Villacorta L, Chang L. The role of perivascular adipose tissue in vasoconstriction, arterial stiffness, and aneurysm. Horm Mol Biol Clin Investig 21: 137–147, 2015. doi:10.1515/hmbci-2014-0048.
    Crossref | PubMed | Web of Science | Google Scholar
  • 135. Wagenseil JE, Mecham RP. Elastin in large artery stiffness and hypertension. J Cardiovasc Transl Res 5: 264–273, 2012. doi:10.1007/s12265-012-9349-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 136. Wang M, Monticone RE, Lakatta EG. Proinflammation of aging central arteries: a mini-review. Gerontology 60: 519–529, 2014. doi:10.1159/000362548.
    Crossref | PubMed | Web of Science | Google Scholar
  • 137. Wang MC, Tsai WC, Chen JY, Huang JJ. Stepwise increase in arterial stiffness corresponding with the stages of chronic kidney disease. Am J Kidney Dis 45: 494–501, 2005. doi:10.1053/j.ajkd.2004.11.011.
    Crossref | PubMed | Web of Science | Google Scholar
  • 138. Weberruß H, Pirzer R, Böhm B, Dalla Pozza R, Netz H, Oberhoffer R. Intima-media thickness and arterial function in obese and non-obese children. BMC Obes 3: 2, 2016. doi:10.1186/s40608-016-0081-9.
    Crossref | PubMed | Google Scholar
  • 139. Weisbrod RM, Shiang T, Al Sayah L, Fry JL, Bajpai S, Reinhart-King CA, Lob HE, Santhanam L, Mitchell G, Cohen RA, Seta F. Arterial stiffening precedes systolic hypertension in diet-induced obesity. Hypertension 62: 1105–1110, 2013. doi:10.1161/HYPERTENSIONAHA.113.01744.
    Crossref | PubMed | Web of Science | Google Scholar
  • 140. Wenzel U, Turner JE, Krebs C, Kurts C, Harrison DG, Ehmke H. Immune mechanisms in arterial hypertension. J Am Soc Nephrol 27: 677–686, 2016. doi:10.1681/ASN.2015050562.
    Crossref | PubMed | Web of Science | Google Scholar
  • 141. Whaley-Connell A, Johnson MS, Sowers JR. Aldosterone: role in the cardiometabolic syndrome and resistant hypertension. Prog Cardiovasc Dis 52: 401–409, 2010. doi:10.1016/j.pcad.2009.12.004.
    Crossref | PubMed | Web of Science | Google Scholar
  • 142. Xia N, Horke S, Habermeier A, Closs EI, Reifenberg G, Gericke A, Mikhed Y, Münzel T, Daiber A, Förstermann U, Li H. Uncoupling of endothelial nitric oxide synthase in perivascular adipose tissue of diet-induced obese mice. Arterioscler Thromb Vasc Biol 36: 78–85, 2016. doi:10.1161/ATVBAHA.115.306263.
    Crossref | PubMed | Web of Science | Google Scholar
  • 143. Yamagishi S, Fukami K, Matsui T. Crosstalk between advanced glycation end products (AGEs)-receptor RAGE axis and dipeptidyl peptidase-4-incretin system in diabetic vascular complications. Cardiovasc Diabetol 14: 2, 2015. doi:10.1186/s12933-015-0176-5.
    Crossref | PubMed | Web of Science | Google Scholar
  • 144. Yamagishi S, Nakamura N, Suematsu M, Kaseda K, Matsui T. Advanced glycation end products: a molecular target for vascular complications in diabetes. Mol Med 21, Suppl 1: S32–S40, 2015. doi:10.2119/molmed.2015.00067.
    Crossref | PubMed | Web of Science | Google Scholar
  • 145. Yen W, Cai B, Yang J, Zhang L, Zeng M, Tarbell JM, Fu BM. Endothelial surface glycocalyx can regulate flow-induced nitric oxide production in microvessels in vivo. PLoS One 10: e0117133, 2015. doi:10.1371/journal.pone.0117133.
    Crossref | PubMed | Web of Science | Google Scholar
  • 146. Young MJ, Rickard AJ. Mineralocorticoid receptors in the heart: lessons from cell-selective transgenic animals. J Endocrinol 224: R1–R13, 2015. doi:10.1530/JOE-14-0471.
    Crossref | PubMed | Web of Science | Google Scholar
  • 147. Yusef YR, Thomas W, Harvey BJ. Estrogen increases ENaC activity via PKCδ signaling in renal cortical collecting duct cells. Physiol Rep 2: e12020, 2014. doi:10.14814/phy2.12020.
    Crossref | PubMed | Google Scholar
  • 148. Zapata-Gonzalez F, Auguet T, Aragonès G, Guiu-Jurado E, Berlanga A, Martinez S, Martí A, Sabench F, Hernandez M, Aguilar C, Sirvent JJ, Jorba R, Del Castillo D, Richart C. Interleukin-17A gene expression in morbidly obese women. Int J Mol Sci 16: 17469–17481, 2015. doi:10.3390/ijms160817469.
    Crossref | PubMed | Web of Science | Google Scholar
  • 149. Zieman SJ, Melenovsky V, Kass DA. Mechanisms, pathophysiology, and therapy of arterial stiffness. Arterioscler Thromb Vasc Biol 25: 932–943, 2005. doi:10.1161/01.ATV.0000160548.78317.29.
    Crossref | PubMed | Web of Science | Google Scholar

AUTHOR NOTES

  • Address for reprint requests and other correspondence: James R. Sowers, University of Missouri, D109 Diabetes Center HSC, One Hospital Dr., Columbia, MO 65212 (e-mail: ).